IES20090074A2 - Listeria monocytogenes cytotoxin listeriolysin S - Google Patents

Listeria monocytogenes cytotoxin listeriolysin S

Info

Publication number
IES20090074A2
IES20090074A2 IES20090074A IES20090074A2 IE S20090074 A2 IES20090074 A2 IE S20090074A2 IE S20090074 A IES20090074 A IE S20090074A IE S20090074 A2 IES20090074 A2 IE S20090074A2
Authority
IE
Ireland
Prior art keywords
cytotoxin
gene
strains
lipi
monocytogenes
Prior art date
Application number
Inventor
Paul Ross
Colin Hill
Paul Cotter
Original Assignee
Univ College Cork Nat Univ Ie
Teagasc Agric Food Dev Authority
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Univ College Cork Nat Univ Ie, Teagasc Agric Food Dev Authority filed Critical Univ College Cork Nat Univ Ie
Priority to IES20090074 priority Critical patent/IES20090074A2/en
Publication of IES20090074A2 publication Critical patent/IES20090074A2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6888Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for detection or identification of organisms
    • C12Q1/689Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for detection or identification of organisms for bacteria
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Analytical Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Wood Science & Technology (AREA)
  • Microbiology (AREA)
  • Immunology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Physics & Mathematics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Medicinal Chemistry (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

The invention relates to a gene derivable from the LIPI-3 pathogenicity island of the gram positive pathogen Listeria inonocyto genes and to the Listeria inonocyto genes cytotoxin, Listeriolysin S. The present invention further relates to the application of Listeriolysin S as a target for diagnostic screening and as a therapeutic agent such as an anti-cancer agent.

Description

The current invention relates to bacterial cytotoxins. More specifically the present invention relates to a Listeria monocytogenes cytotoxin, Listeriolysin S. The present invention further relates to the application of Listeriolysin S as a target for diagnostic screening and as a therapeutic agent such as an anti-cancer agent.
Background to the Invention listeria monocytogenes is a food-borne gram-positive pathogenic bacterium responsible for listeriosis, a life-threatening infection of pregnant and immuno-compromised humans. Strains of L. monocytogenes can be separated into three evolutionary lineages (I-IIl), ail of which are capable of causing disease in humans and animals.
Uneage i-lineage 111 all possess the well-established virulence genes associated with the pathogenesis and intracellular life cycle of L. monocytogenes. These genes are located on a 9-kb virulence gene island designated Listeria monocytogenes Pathogenicity Island 1 (I J PI-1). U1P1-1 contains six genes, all of which are directly involved in pathogenesis.
The gene hly located on LIPI-1 encodes Listeriolysin 0 (LLO), a pore forming thiolactivated toxin secreted by L. monocytogenes. The intracellular life cycle of A. monocytogenes begins with adhesion and subsequent penetration into a host eurkaryotic cell. At this point during the initial entry into the cell the bacterium becomes engulfed by a host vacuole. The disruption and escape from the host vacuole by the bacterium is essential for intracellular survival and proliferation. It is at this point that LLO is considered to play an essential role, facilitating the survival of the bacterium in an infected cell. All L.monocytogenes strains, regardless of lineage, produce Listeriolysin 0. It is well known in the art that lineage 1 strains (consisting primarily of strains of serotype 1 /2b and 4b) are responsible for the vast majority of outbreaks of listeriosis and almost two thirds of human sporadic cases. This lineage includes three previously defined epidemic clones responsible for multiple listeriosis outbreaks in Europe and North America.
However, the genetic basis for the enhanced virulence of lineage I strains has not been established. It is not possible to reliably discriminate between epidemic” and non- Ο ο 74 epidemic” strains of the disease in animal models. Furthermore, on the basis that there have been a few exceptional outbreaks by non-lineage I strains and that these stains are also responsible for the remaining third of sporadic cases, it is evident that should a lineage I-specific factor exist its presence is not a pre-requisite for pathogenesis but rather an indication of a higher virulence potential.
A previous study by Nelson et al revealed there to be relatively little genetic difference between the high and low virulence strains of L. monocytogenes. For example, there are a mere 51 genes present in serotype 4b strains (F2365, H7858; lineage I), which are lacking from their 1 /2a (EGDe, F6854; lineage 11) counterparts. To date, however, no candidate genes have been proposed which could explain the enhanced virulence of lineage I L.monocytogenes strains, A variety of laboratory procedures have been developed in an attempt to accurately determine the subtype of L. monocytogenes strains. These methods have been principally serological (e.g. Listeria serotyping antiserum SEIKEN; Seiken Co. Ltd., Tokyo, Japan) and nucleic acid based techniques including PCR (e.g. multiplex PCR can discriminate between strains from different lineages (/)) DNA sequencing (e.g. multi-locus sequence typing (MLST) has been used to differentiate between and within lineages (2-4), hybridization (e.g. single nucleotide polymorphism (SNP)-typing has been used to identify different sequence types and multilocus genotypes (MLGTs) within specific lineages (5)), pulse field gel electrophoresis (e.g. to differentiate between different patterns among 4b strains (6)) and ribotyping (e.g. subtyping within lineages (7, 8)). Although these methods have proven successful at differentiating between the or within different lineages of L. monocytogenes, they are not effective at determining the virulence potential of L. monocytogenes strains as highly virulent strains can be found within each lineage.
The identification and differentiation of L.monocytogenes strains is a key problem facing the food and medical industries. Therefore, the availability of a diagnostic kit with the potential to differentiate between high and low virulence strains would be of great benefit to food producers, vets and clinicians. This in tandem will allow legislation to adjust the levels of L. monocytogeneous that are permitted in food in order to effectively control levels of L. monocytogenes and hence to prevent infection of listeriosis.
The Object of the Invention It is an object of the current invention to provide a cytotoxic agent, which can be used as a therapeutic agent. Another object is to provide a lineage I specific cytolytic virulence factor, called listeriolysin (Lis) S and its use as a target for diagnostic screening. Lis detection can be used to distinguish between high and low virulence L. monocytogenes strains. It is a further object of the invention to provide a therapeutic agent for the treatment of disease. Lis or bacteria producing Lis are cytotoxic to a number of cell types including cancerous cells. Lis has an application as a chemotherapeutic agent, potentially acting as an alternative to existing agents e.g. bleomycin (treatment of Hodgkin lymphoma, squamous cell carcinomas, and testicular cancer, pleuodesis and plantar warts) and epothilone (a novel anti-cancer peptide currently undergoing clinical trial).
Summary of the Invention According to the present invention there is provided a gene derivable from the LIPI-3 pathogenicity island of the gram positive pathogen Listeria monocytogenes, the island being flanked by two Rho-independent terminators, located after Imof2365_l 111 and ilsP, respectively, and two Rho-independent duplicated glyoxalase-encoding genes (Imof2365 1111,1121). The island is downstream of lmof2365_l 110 and upstream of lmof2365_1127. The location of these genes is significant as corresponding genes have been identified at a similar location within the genomes of all L. monocytogenes sequenced to date. The gene may be selected from the genes llsA, 1 IsG, llsH, llsX, llsB, llsY, llsD and IlsP. The gene may be associated with a subset of lineage I strains of Listeria monocytogenes.
The current invention also provides a nucleic acid sequence having the sequence of SEQ ID No. 1 or a fragment of SEQ ID No. 1, or a sequence substantially similar thereto also encoding a cytotoxic agent. The invention also provides an amino acid sequence corresponding to the above nucleic acid sequence having SEQ ID No, 2 or a sequence substantially similar thereto also encoding a cytotoxic agent.
SEQ ID No. 1: ATGAATATTAAATCACAATCATCAAATGGCTACAGTAATAATGCTGTAGGCT CTGAAGCAATGAACTATGCAGCTGGATGTTGCTCATGTTCTTGTTCAACTTGC ACATGTACATGTACATGCGCATCATCTGCTGCAACCAAAATGTAA SEQ ID No.2: MNIKSQSSNGYSNNAVGSEAMNYAAGCCSCSTCTCTCTCASSAATKM.
The sequence encodes a cytotoxin. The nucleic acid sequence is located on a pathogenicity island of the gram-positive pathogen Listeria monocytogenes referred to as LIPI-3. LIPI-3 contains the 8 genes HsA, llsG, llsH, llsX, llsB, llsY, llsD and llsP. The 8gene region is flanked by two Rho-independent terminators, located after Imof2365_1111 and llsP, respectively, and two Rho-independent duplicated glyoxalase-encoding genes (Imof2365Jlll, J121). The outer limits of the LIPI-3 are also apparent from comparison with the genome sequences of strains that lack the island i.e. a number of these strains possess genes corresponding lmof2365_llll and 1121 but none possess any of the Ils genes. LIPI-3 is also distinguished by having a very high genomic dissimilarity value (δ* ofO.l 178) relative to that of the remainder of the F2365 genome (δ* of 0.0343) and an atypical %GC content (29.9 vs 38%).
The invention also provides a nucleic acid sequence selected from SEQ ID NO. 3,4, 5 7, 9, 11, 13,15, 17and 19 and an amino-acid sequence selected from SEQ ID NO 6, 8, 10, 12, 14, 16,18, and 20 and sequences which are substantially similar thereto.
By ‘substantially similar thereto’ is meant sequences which are homologous to the specified sequences, particularly sequences which are about 70% homologous under low stringency conditions, or about 80% homologous under medium stringency conditions, or about 90% or about 95% homologous thereto under high stringency conditions. ‘Stringency” is used in reference to the conditions of temperature, ionic strength, and the presence of other compounds such as organic solvents, under which nucleic acid hybridizations are conducted. Those skilled in the art will recognize that “stringency” conditions may be altered by varying the parameters just described either individually or in concert. With “high stringency” conditions, nucleic acid base pairing will occur only between nucleic acid fragments that have a high frequency of complementary base sequences (for example, hybridization under “high stringency” conditions, may occur between homologs with about 85-100% identity, preferably about 70-100% identity). With medium stringency conditions, nucleic acid base pairing will occur between nucleic acids with an intermediate frequency of complementary base sequences (for example, hybridization under “medium stringency” conditions may occur between homologs with IE Ο 9 Ο 0 74 I* about 50-70% identity). Thus, conditions of “weak” or “low” stringency are often required with nucleic acids that are derived from organisms that are genetically diverse, as the frequency of complementary sequences is usually less.
“High stringency conditions” when used in reference to nucleic acid hybridization comprise conditions equivalent to binding or hybridization at 42° C. in a solution consisting of 5xSSPE (43.8g/l NaCl, 6.9 g/l NaH2PO4H2O and 1.85 g/l EDTA, ph adjusted to 7.4 with NaOH), 0.5% SDS, 5xDenhardt’s reagent and 100pg/ml denatured salmon sperm DNA followed by washing in a solution comprising O.lxSSPE, 1.0%SDS at 42° C. when a probe of about 500 nucleotides in length is employed.
“Medium stringency conditions” when used in reference to nucleic acid hybridization comprise conditions equivalent to binding or hybridization at 42° C. in a solution consisting of 5Xsspe (43.8 g/l NaCl, 6.9 g/l NaH2PO4H2O and 1.85 g/l EDTA, pH adjusted to 7.4 with NaOH), 0.5% SDS, 5xDenhardt’s reagent and 100 pg/ml denatured salmon sperm DNA followed by washing in a solution comprising l.OxSSPE, 1.0% SDS at 42° C, when a probe of about 500 nucleotides in length is employed.
‘Low stringency conditions” comprise conditions equivalent to binding or hybridization at 42° C. in a solution consisting of 5xSSPE (43.8 g/l NaCl, 6.9 g/l NaH2PO4H2O and 1,85 g/l EDTA, pH adjusted to 7.4 with NaOH), 0.1% SDS, 5xDenhardt’s reagent [50xDenhardt’s contains per 500ml: 5g Ficoll (Type 400, Pharamcia), 5 g BSA (Fraction V; Sigma)] and 100 pg/ml denatured salmon sperm DNA followed by washing in a solution comprising 5xSSPE, 0.1% SDS at 42° C, when a probe of about 500 nucleotides in length is employed.
SEQ ID NO 3; LIPI-3 GTGGAGTGAAATATAAGTTAGAGATTATTTTTCGATTAGGTGTACTTTTTTTTT GTCTTCGTTTTATAGATTTTAGTATTTTTTTAATCTTGTATTATTAATGATAGTT ATCTATAGCTTATATGCAGATTATGGAGTTGTATTAGTAAAAAAGCCCTGAA GCCTCAGCATTATTGAGTTTCTAGGGCTTTTTTATGTCGAGTGATAACCGTTTT TTATTATCGTACTATTTTTGTGGATAATATTTTGTCCAATTATACATATATACT TTTGGAGATATTTAAACCAATTTTGCATATTATCAAACGGAGGGATATATTTT TTATAAAAATAATTAAAAAAATTATTTAATTTTCTGAAATAAACAAAGAATTT IE Ο 9 ο Ο 74 ATTTATTTTGATAATTATATTGAAAACGATTTCACAATGTGATAGGATGAACT AAGGGATAATTTATTCCAAAAAATAAAAAGGAGGCATTTGAATGAATATTAA ATCACAATCATCAAATGGCTACAGTAATAATGCTGTAGGCTCTGAAGCAATG AACTATGCAGCTGGATGTTGCTCATGTTCTTGTTCAACTTGCACATGTACATG TACATGCGCATCATCTGCTGCAACCAAAATGTAAATTTTAGAATGAAAAAGG GATACATTTGTGTCCCTTTTCAATAAGTTATGAGGTGCATGTGCATATGAATA ATATTTTGGAAACGAAAAATTTGAAAGTTACAATAAATAATAAAGTAATTCT ATATTTAGATAAAGAAGTTTGTATTTCAGAAAAAGACAAAGTTGCCATTCTA GGAGACAATGGAGCTGGGAAAACCACGTTAGTAAATAGTATTCTTGGTGAAA AGAACTCTTCTGGAGAAATTACCAAAAAGTTTAAAAAAAATGACTGTGGTGT AGTGTTTCAAGAAAATGCATATAATGATTTGATGAAAGTTTATGAATTGATTA CTTTAGTTCTGCCACATCTAAAGAAAAAAGAGAGAGCGCAGTTTTTACACAA ATATGAACTTGAAAGTTTGAGAAAAAAGTACATTAAAGACTTATCTGGTGGA GAAAAACAACGACTAACACTATGTTTAGTACTAGAAAGTCATAAGAAATTAT ATATCTTTGATGAGTTAACTTCAGGATTAGATTATAAGAAGAGACTAGGTTTA CTTGCTTTGATGAAAGAAAAAACTAAGGACGCGACGGTGATAAATATAACGC ATTACTTTGAGGAAGTTGAAAACTGGGCAACGAAAGTTTTGATTCTTCAAAA AGGAATTTTACTTTTTTGGGGAACAATATCAGAATTTTTCTCTAATTTTCCTCA TTATTCTGTTATTAAAGTAGATCAAATCGAACTAACAAAGATAGATGAAACA GATATGACTTTTATGCAAAGCACAGATACAGGTGACGGGATAGCAGTTATTT GTTCGGATTTGCAAATTCAGGAAGAGACAAAGAAAATTTTGGATAAAAAAAA CGTCACATACAACACAATAAAACAAAATATATATACAACTTATTTAGTTGCA TATTTGCGCGGAACATCAAGCAGTGAACAGGAGGTACTGATATAATGAGTTA TTTATGGACAAGTATTAAAATGCAATTTAGAATTCCAGTATCTGTGTTTTTCT CATTATTATTTCCACTAATCATGATGTTCGCTATGGTTACTTCTTATGGAAACT TTGATATTGGTGAAGGTTATCATTTTGTAGATAAATATTTTTTGATTAGTACA GGGATGGGCATGTTACCAATAGCATTAAyqAGTTTTCCAATCTGGCTTGGTGA AAGTGTTCAAAATAAAAGCTATAAAAGATTAGAATATTTTGGATTGAGTGCA CAGAAAATAATTGTTTCAGATGTATGTTCTTATATTTTGCTAACAGCCTTAAG TATTTTTGTCAATATACTGTTTGGCTACTTGGTATATGGACTACATATCCCAG ACTGGCAATATTTTATTGCTTATGTTTTGCAATGTCTGTATTGTAATCTAGTGC IE Ο 9 0 g TTTTGATTTTTGGTGCCTTGCTTGCTTTAATTATTAAAAATCCCCGGATATTGA TGCCAGTAGGAATGTGTTTATTATTTATGTTTTATATTTTTACAGGAACATTTT CATCTTTTTCGGAACTTCCTAAATCATTTCAAGCAGTTGGTAATTTTTTACCTA TGAAATATATAATGAATGATTTATTTAATpTTTGGACACAAAATAAATTGTTC ATCTCTAAGTTTTTAGCTTTAAACACGTTATATGGAATCATCTTATCATTCGCA CTAGTTGCTTTTTTGTTGAGGCAAAAGAAAATAAAAATTAAACCATAATTTAT TATACTCAAGGTATAGAAAGGATTGTTTGTAATCTATGAAAAAAAAATTTAG TAATCCCACGTTTAGAATTATTGCATCAATTGTTCTAGGGATTTTGATAGGTG TTCTTATTTGCTTTGTGGCTATTGGTTTAGGTTACATTCACATGAATGATGGCA CATTAAAAGAGTATAGTGTGAAAATTTTCGGACTAACTATTTTTGATATTAAA AGAGTTGGTAGTGAGATGGTGGGAACACCTAATAACACTAGCATGATGTTTA TAGGGGTTATTATTTCTATGATACTAGCTATTGTTGTAGAAATTATTGTTTCGT TAAAAAACAGACATAGAAAGGAAACAGCAAAATGATCGACTATGAGAAAAA AGGCTTTTTTAACATCCACACATTGGTAAATAAAGATAATGCTAATATTTCTA ATAGTGATAATAAACATATTTATTCCCAGCTGATGTCTGGTAATGGCAATTCA CCAATGCTAGGTTATTTATTAAATATGAATAAACAAAACTTGAATGACTTCAA AAGTATCATGTTTTATAATGAATCTAATCTAGCTTCTTTAATTAACGAGGCAA ,· I GAGAAATGGAAGAATTAATAGATAGCTCAACTCTTTTTTTAAGTAAAACAAA TAAAAAAATTAAAACCCATTTTTCTAAAGTGCTTGAGCAAAGACATAGTACA AGAAATTTTGTATACGAAACCATGGATTTATCAACATTCTCAAATATCATACA GTTTTCTTTTGGCCTTAGTACAAGAAAGTTGGTTTATAACGACTTACAATCAA CCACCAGGCATTATTCTTCTGGGGGCGGTTTATATCCAATTGATGTTTTTTTGT ATATAAATAATATTTCTGGAATTGCCAAGGGTATATATAAATATCAGCCATAC ACGCATAGCTTGCATCCTCTAGATGTAGATAAAATTGATGTAGAGTCGTTTTT CGTAGGTGATAATATTGACACTTCTAATATGAATTTTTGTGTTTTCTTTGGATA CTCTATTAATAAAAACTATGTGAAATATGGAGAACTATCTTTACTGAATACGT TTGTGGAACTAGGAGGAATATCACACAACTTTGATTTAGTTTGTCATTCGGTT CACTATACAAGTTGTCCTATTGCAGGTTTTAACAAGTCGTACATAGAGAAGCT TTTATATTTAGATGGGATAAATGATCACATTATATTTACAAATATTTGTGGAA AGGAATGATATTTTTATGAAAAATTTTGATATTAGAATAGGAACGCAGACAA TTGACAATGATACGGAATTCATTTTAAAAAGAGGGGTTATTCATAAGAATGA IE Ο 9 ο ΰ 7 4 GATCGTTATCAATAAAGAGGAAAGTTCAAAAGAATTTGTTTCAACATTTAAA GAATTGGTCAAGAAAAAAACTATTACTATATCTTCAGAAGATGCAATCTACA ATGATTTTGAAACACTTACAAAATTTGGCTTTTTAACAATTTCTAAAAACCAA ACCTTAAAACCTCTCTTGATAGTTGAAGATGCTTTGTTTGATGACGTGAAGAG CTATTTCCAGGAAGAAATTGAGATTTTATCATCCTCTGAATTTCTTTTAAAAA AAGATATTCGGTTACTTACTGAGAACAAAGATATATTGCAACTTACTAAACT AGTAGATGAAAAAAAGGAATTAATGAAAAATTATAATTATATTTATTTAATC ACAAACATTTCCAATATTTCCTTATTACGTGGATTTAACAAGTTAATGAAAGA GACTAACAGTGTTAATACCATCGCATTTTTTGATAATGAAAATGTATTTGTTA CTTGTATAGAACATGGAGAAACTGGCTGCTATGAGTGTTTAGAGCGGCAACT ACTCTCTCATTTTGATGGTGTTGTTACGGATTATTTAGTACAGTCAGAAAATA ATGTTTCCACAGCTGAATTGATGTTTGTTCTTTCAATCATAAAAAAAGAAATC GAAAATACCTCTATTTATGGTCAATCTTCTCTGTTAGGCAATCTTCTCCACTTT AATTTTAATAACTACGAGTATACGTTTAATACAAATAGAATCCAAAGTTGTTG TTCTACCTGTGCTACTTTCAATAATATATTATTTGAAGAGCAAAACATTCGGT CAGTGAATATACTAAAGGAGCTGATGAGTAGTGATTAGCATACAAAATAATT TGGAATATAACAAGTTACGTTGGGAAACTTTGAGTGGAAATGTAACAGGAAT ATGGGAGAATAATAAGTTCTTTCTAGGCTCTAGTTCTTACCCTATTATGAAGT ATCATTACATTACAGCTAATTTTGTTAATTTTGAAAAACATATTTCTGAAAAT ATGCCTAAAATAAGTTACCATTTAAGTGGATATGGTGTTAATTTTAATGAAGC GCTTGTTAGTTTTATTGGTGAAAGCGCAGAGAGATATACATATTCCTTACTAC CTACTATTATTAAGGACAGGATTATTTTTAGATCATATGAAGAAATGACAAA AGAATATAAAACTGACTTAATATGTGAACTTAAATACATAAATTCGTATTACT CTTCTGAAGTATGCGAAAATTATGTTACTCCAAATGATACTATTCAATGGATA GCAATGAATTCTCTTGTTCATTCTGATAAAAAAGTATGGATGCCTTTGCAATT TGTTACTATGTATACAGAAGAAATGTTTTCTAATGAAAAAAGATATGTAACT AGCGCTGTATCAACAGGCACTGCTTGTCATGAAACTGTAGAAAAAAGTATAG AAAATGCCCTAATTGAATATCTTCAAATTGATTCCTTTAATTTATGGTGGTAT GGAGGGTTTCGTGCGAGAGATATAGAAATAGATATCACTCGAAATATATCAA GCTGGTTCGACAATCAAGTAGCTGTGAAAAAATTTTTATCAAAGTTTAATGTA CATTTTTCAGATATAAGTTTTGATAAATCAATTTATATTGTGCTATGCGAAAT Il<“> 0»7( AGAGGCAAAAAATTCAAGTGATGCCTTTCCTAAATATACTGTTGGCGTTCAA GGCGGATATTCACTAGATAAATCAATATATCGTGCATTTATGGAATGTCTAAC TGTACTAGAATATAATATGAATGTCACTTGGACAGATAAAGAGAAATTTCTTT CAGTCACACAGGAAACACGTGTTATAGATAATTTGGATGATAATGTTATTTAT TATTCAAAGTATGGAAAACCAGAATTGCAATATAATACTAATCAATTAAAGA atgatacggaaaaagttacaaatctgaaggcacttttagaaaagttgcccac AATTAGTCAGTATGCTGCATTCTTACCTATCACACCTTCAGAGTTTAGATATA TGAATTGTGAAATAAGCAGGGTGATTCTTCCAGAATTGTTATCCATTCATCTT CCTTCATATCCGCCTTACTACCATGTAAGATATGAAGAAATTGGAGGTGTGGT AAATAATATTCCACATCCAATTGCATAACATCTTTTTTTTAATCACATTATTTC CAGGTATGCTTCTTTTATTGACGAAATGGATTCCAGTTCTATCTAGAAAGAGT ACTTTTTTTCAATATTTACTTTGTTTATTCTTGATCACAATTATGAATAGTTTG TTTTTCCGTCAACAGTTTGTGGTAGTTTTATCGCTGATATGTATTTTATTCTTA CCATTTATTCTGTTTTTTGTAGAATATATATTTGTTGAGAGACAATGGAAAAA GTTGCTTACTATTTATAAAAAAAATAAAATTTTTATCCAATCTATTGTATGGT TTCCTGTTTTAGAAGAAATAATTTTCCGTTTTTTTATTTATCAATACTGTGAGT TATTTGATTTCAGTAATATCCAGTATATATTACTAGCCACCTTTTCATTCGTGA TTGCACATATTTTTTATCAAGGAGTGTCTTCAATTGTCAAAATACTATTTTCTT TTATATTAAGTATATTATTTTTATTAACACTAAATATATTTTTGACAATAATAA TTCACTGTATTTTCAACTTTTTAGTTTATATAGTTCGTACTAGTAAATATGAGA ACCACCGTAATTGGTAAGCTGACAGTATTATATTAATTTAGCTGTTAAAGTAT GGAAATACCAATAAATCAACGTTTTTCAACTTATGAAATGCTTGGTTCTCATT AAAAATGATATATTTCCCTACCA SEQ ID NO 4;Promoter PllsA GTGGAGTGAAATATAAGTTAGAGATTATTTTTCGATTAGGTGTACTTTTTTTTT GTCTTCGTTTTATAGATTTTAGTATTTTTTTAATCTTGTATTATTAATGATAGTT ATCTATAGCTTATATGCAGATTATGGAGTTGTATTAGTAAAAAAGCCCTGAA GCCTCAGCATTATTGAGTTTCTAGGGCTTTTTTATGTCGAGTGATAACCGTTTT TTATTATCGTACTATTTTTGTGGATAATATTTTGTCCAATTATACATATATACT TTTGGAGATATTTAAACCAATTTTGCATATTATCAAACGGAGGGATATATTTT TTATAAAAATAATTAAAAAAATTATTTAATTTTCTGAAATAAACAAAGAATTT ,Ε Ο 0 74 ATTTATTTTGATAATTATATTGAAAACGATTTCACAATGTGATAGGATGAACT AAGGGATAATTTATTCCAAAAAATAAAAAGGAGGCATTTGA SEQ ID NO 5; /kA ATGAATATTAAATCACAATCATCAAATGGCTACAGTAATAATGCTGTAGGCT CTGAAGCAATGAACTATGCAGCTGGATGTTGCTCATGTTCTTGTTCAACTTGC ACATGTACATGTACATGCGCATCATCTGCTGCAACCAAAATGTAA (Predicted leader peptide-encoding region in standard text, predicted unmodified leaderless propeptide-encoding region in bold text) SEQ ID NO 6; Unmodified LlsA MN1KSQSSNGYSNNAVGSEAMNYAAGCCSCSTCTCTCTCASSAATKM (Predicted leader peptide in standard text, predicted unmodified leaderless propeptide in bold text) SEQ ID NO 7; llsG ATGAATAATATTTTGGAAACGAAAAATTTGAAAGTTACAATAAATAATAAAG TAATTCTATATTTAGATAAAGAAGTTTGTATTTCAGAAAAAGACAAAGTTGCC ATTCTAGGAGACAATGGAGCTGGGAAAACCACGTTAGTAAATAGTATTCTTG GTGAAAAGAACTCTTCTGGAGAAATTACCAAAAAGTTTAAAAAAAATGACTG TGGTGTAGTGTTTCAAGAAAATGCATATAATGATTTGATGAAAGTTTATGAAT TGATTACTTTAGTTCTGCCACATCTAAAGAAAAAAGAGAGAGCGCAGTTTTT ACACAAATATGAACTTGAAAGTTTGAGAAAAAAGTACATTAAAGACTTATCT GGTGGAGAAAAACAACGACTAACACTATGTTTAGTACTAGAAAGTCATAAGA AATTATATATCTTTGATGAGTTAACTTCAGGATTAGATTATAAGAAGAGACTA GGTTTACTTGCTTTGATGAAAGAAAAAACTAAGGACGCGACGGTGATAAATA TAACGCATTACTTTGAGGAAGTTGAAAACTGGGCAACGAAAGTTTTGATTCTT CAAAAAGGAATTTTACTTTTTTGGGGAACAATATCAGAATTTTTCTCTAATTT TCCTCATTATTCTGTTATTAAAGTAGATCAAATCGAACTAACAAAGATAGATG AAACAGATATGACTTTTATGCAAAGCACAGATACAGGTGACGGGATAGCAGT TATTTGTTCGGATTTGCAAATTCAGGAAGAGACAAAGAAAATTTTGGATAAA AAAAACGTCACATACAACACAATAAAACAAAATATATATACAACTTATTTAG TTGCATATTTGCGCGGAACATCAAGCAGTGAACAGGAGGTACTGATATAAT IE09q 0 74 SEQ ID NO 8; LIsG MNNILETKNLKVTINNKVILYLDKEVCISEKDKVA1LGDNGAGKTTLVNSILGEK NSSGEITKKFKKNDCGVVFQENAYNDLMKVYELITLVLPHLKKKERAQFLHKYE LESLRKKY1KDLSGGEKQRLTLCLVLESHKKLYIFDELTSGLDYKKRLGLLALMK EKTKDATVINITHYFEEVENWATKVL1LQKGILLFWGTISEFFSNFPHYSV1KVDQI ELTKIDETDMTFMQSTDTGDGIAVICSDLQ1QEETKKILDKKNVTYNT1KQN1YTT YLVAYLRGTSSSEQEVLI SEQ ID NO 9; ilsff ATGAGTTATTTATGGACAAGTATTAAAATGCAATTTAGAATTCCAGTATCTGT GTTTTTCTCATTATTATTTCCACTAATCATGATGTTCGCTATGGTTACTTCTTA TGGAAACTTTGATATTGGTGAAGGTTATCATTTTGTAGATAAATATTTTTTGA TTAGTACAGGGATGGGCATGTTACCAATAGCATTAATCAGTTTTCCAATCTGG CTTGGTGAAAGTGTTCAAAATAAAAGCTATAAAAGATTAGAATATTTTGGAT TGAGTGCACAGAAAATAATTGTTTCAGATGTATGTTCTTATATTTTGCTAACA GCCTTAAGTATTTTTGTCAATATACTGTTTGGCTACTTGGTATATGGACTACAT ATCCCAGACTGGCAATATTTTATTGCTTATGTTTTGCAATGTCTGTATTGTAAT CTAGTGCTTTTGATTTTTGGTGCCTTGCTTGCTTTAATTATTAAAAATCCCCGG ATATTGATGCCAGTAGGAATGTGTTTATTATTTATGTTTTATATTTTTACAGGA ACATTTTCATCTTTTTCGGAACTTCCTAAATCATTTCAAGCAGTTGGTAATTTT TTACCTATGAAATATATAATGAATGATTTATTTAATGTTTGGACACAAAATAA ATTGTTCATCTCTAAGTTTTTAGCTTTAAACACGTTATATGGAATCATCTTATC ATTCGCACTAGTTGCTTTTTTGTTGAGGCAAAAGAAAATAAAAATTAAACCAT AA SEQ ID NO 10; LIsH MSYLWTSIKMQFRIPVSVFFSLLFPLIMMFAMVTSYGNFD1GEGYHFVDKYFLIST GMGMLP1ALISFPIWLGESVQNKSYKRLEYFGLSAQKIIVSDVCSYILLTALS1FVNI LFGYLVYGLHIPDWQYFIAYVLQCLYCNLVLLIFGALLALIIKNPRILMPVGMCLL FMFYIFTGTFSSFSELPKSFQAVGNFLPMKYIMNDLFNVWTQNKLFISKFLALNTL YGIILSFALVAFLLRQKKIKIKP SEQ ID NO 11; llsX ,£ 0 9 0 Ο 74 ATGAAAAAAAAATTTAGTAATCCCACGTTTAGAATTATTGCATCAATTGTTCT AGGGATTTTGATAGGTGTTCTTATTTGCTTTGTGGCTATTGGTTTAGGTTACAT TCACATGAATGATGGCACATTAAAAGAGTATAGTGTGAAAATTTTCGGACTA ACTATTTTTGATATTAAAAGAGTTGGTAGTGAGATGGTGGGAACACCTAATA ACACTAGCATGATGTTTATAGGGGTTATTATTTCTATGATACTAGCTATTGTT GTAGAAATTATTGTTTCGTTAAAAAACAGACATAGAAAGGAAACAGCAAAAT GA SEQ ID NO 12; LlsX MKKKFSNPTFRIIASIVLGILIGVL1CFVAIGLGYIHMNDGTLKEYSVKIFGLTIFDIK RVGSEMVGTPNNTSMMFIGVIISMILAIVVEIIVSLKNRHRKETAK SEQ ID NO 13; llsB ATGATCGACTATGAGAAAAAAGGCTTTTTTAACATCCACACATTGGTAAATA AAGATAATGCTAATATTTCTAATAGTGATAATAAACATATTTATTCCCAGCTG ATGTCTGGTAATGGCAATTCACCAATGCTAGGTTATTTATTAAATATGAATAA ACAAAACTTGAATGACTTCAAAAGTATCATGTTTTATAATGAATCTAATCTAG CTTCTTTAATTAACGAGGCAAGAGAAATGGAAGAATTAATAGATAGCTCAAC TCTTTTTTTAAGTAAAACAAATAAAAAAATTAAAACCCATTTTTCTAAAGTGC TTGAGCAAAGACATAGTACAAGAAATTTTGTATACGAAACCATGGATTTATC AACATTCTCAAATATCATACAGTTTTCTTTTGGCCTTAGTACAAGAAAGTTGG TTTATAACGACTTACAATCAACCACCAGGCATTATTCTTCTGGGGGCGGTTTA TATCCAATTGATGTTTTTTTGTATATAAATAATATTTCTGGAATTGCCAAGGG TATATATAAATATCAGCCATACACGCATAGCTTGCATCCTCTAGATGTAGATA AAATTGATGTAGAGTCGTTTTTCGTAGGTGATAATATTGACACTTCTAATATG AATTTTTGTGTTTTCTTTGGATACTCTATTAATAAAAACTATGTGAAATATGG AGAACTATCTTTACTGAATACGTTTGTGGAACTAGGAGGAATATCACACAAC TTTGATTTAGTTTGTCATTCGGTTCACTATACAAGTTGTCCTATTGCAGGTTTT AACAAGTCGTACATAGAGAAGCTTTTATATTTAGATGGGATAAATGATCACA TTATATTTACAAATATTTGTGGAAAGGAATGA SEQ ID NO 14; LlsB MIDYEKKGFFNIHTLVNKDNANISNSDNKHIYSQLMSGNGNSPMLGYLLNMNKQ NLNDFKSIMFYNESNLASLINEAREMEELIDSSTLFLSKTNKKIKTHFSKVLEQRH IE 0 9 STRNFVYETMDLSTFSNIIQFSFGLSTRKLVYNDLQSTTRHYSSGGGLYPIDVFLYI NNISGIAKGIYKYQPYTHSLHPLDVDKIDVESFFVGDNIDTSNMNFCVFFGYSINK NYVKYGELSLLNTFVELGGISHNFDLVCHSVHYTSCPIAGFNKSYIEKLLYLDGIN DHI1FTNICGKE SEQ ID NO 15; llsY ATGAAAAATTTTGATATTAGAATAGGAACGCAGACAATTGACAATGATACGG AATTCATTTTAAAAAGAGGGGTTATTCATAAGAATGAGATCGTTATCAATAA AGAGGAAAGTTCAAAAGAATTTGTTTCAACATTTAAAGAATTGGTCAAGAAA AAAACTATTACTATATCTTCAGAAGATGCAATCTACAATGATTTTGAAACACT tacaaaatttggctttttaacaatttctaaaaaccaaaccttaaaacctctct TGATAGTTGAAGATGCTTTGTTTGATGACGTGAAGAGCTATTTCCAGGAAGA AATTGAGATTTTATCATCCTCTGAATTTCTTTTAAAAAAAGATATTCGGTTAC TTACTGAGAACAAAGATATATTGCAACTTACTAAACTAGTAGATGAAAAAAA GGAATTAATGAAAAATTATAATTATATTTATTTAATCACAAACATTTCCAATA TTTCCTTATTACGTGGATTTAACAAGTTAATGAAAGAGACTAACAGTGTTAAT ACCATCGCATTTTTTGATAATGAAAATGTATTTGTTACTTGTATAGAACATGG AGAAACTGGCTGCTATGAGTGTTTAGAGCGGCAACTACTCTCTCATTTTGATG GTGTTGTTACGGATTATTTAGTACAGTCAGAAAATAATGTTTCCACAGCTGAA TTGATGTTTGTTCTTTCAATCATAAAAAAAGAAATCGAAAATACCTCTATTTA TGGTCAATCTTCTCTGTTAGGCAATCTTCTCCACTTTAATTTTAATAACTACGA GTATACGTTTAATACAAATAGAATCCAAAGTTGTTGTTCTACCTGTGCTACTT TCAATAATATATTATTTGAAGAGCAAAACATTCGGTCAGTGAATATACTAAA GGAGCTGATGAGTAGTGATTAG SEQ ID NO 16; LlsY MKNFDIRIGTQTIDNDTEFILKRGVIHKNEIVINKEESSKEFVSTFKELVKKKTITIS SEDAIYNDFETLTKFGFLTISKNQTLKPLLIVEDALFDDVKSYFQEEIEILSSSEFLL KKDIRLLTENKDILQLTKLVDEKKELMKNYNYIYLITNISNISLLRGFNKLMKETN SVNT1AFFDNENVFVTCIEHGETGCYECLERQLLSHFDGVVTDYLVQSENNVSTA ELMFVLS11KKE1ENTSIYGQSSLLGNLLHFNFNNYEYTFNTNRIQSCCSTCATFNNI LFEEQNIRSVNILKELMSSD SEQ ID NO 17; llsD GTGATTAGCATACAAAATAATTTGGAATATAACAAGTTACGTTGGGAAACTT TGAGTGGAAATGTAACAGGAATATGGGAGAATAATAAGTTCTTTCTAGGCTC TAGTTCTTACCCTATTATGAAGTATCATTACATTACAGCTAATTTTGTTAATTT TGAAAAACATATTTCTGAAAATATGCCTAAAATAAGTTACCATTTAAGTGGA TATGGTGTTAATTTTAATGAAGCGCTTGTTAGTTTTATTGGTGAAAGCGCAGA GAGATATACATATTCCTTACTACCTACTATTATTAAGGACAGGATTATTTTTA GATCATATGAAGAAATGACAAAAGAATATAAAACTGACTTAATATGTGAACT TAAATACATAAATTCGTATTACTCTTCTGAAGTATGCGAAAATTATGTTACTC CAAATGATACTATTCAATGGATAGCAATGAATTCTCTTGTTCATTCTGATAAA AAAGTATGGATGCCTTTGCAATTTGTTACTATGTATACAGAAGAAATGTTTTC TAATGAAAAAAGATATGTAACTAGCGCTGTATCAACAGGCACTGCTTGTCAT GAAACTGTAGAAAAAAGTATAGAAAATGCCCTAATTGAATATCTTCAAATTG ATTCCTTTAATTTATGGTGGTATGGAGGGTTTCGTGCGAGAGATATAGAAATA GATATCACTCGAAATATATCAAGCTGGTTQGACAATCAAGTAGCTGTGAAAA AATTTTTATCAAAGTTTAATGTACATTTTTCAGATATAAGTTTTGATAAATCA ATTTATATTGTGCTATGCGAAATAGAGGCAAAAAATTCAAGTGATGCCTTTCC TAAATATACTGTTGGCGTTCAAGGCGGATATTCACTAGATAAATCAATATATC GTGCATTTATGGAATGTCTAACTGTACTAGAATATAATATGAATGTCACTTGG ACAGATAAAGAGAAATTTCTTTCAGTCACACAGGAAACACGTGTTATAGATA ATTTGGATGATAATGTTATTTATTATTCAAAGTATGGAAAACCAGAATTGCAA TATAATACTAATCAATTAAAGAATGATACGGAAAAAGTTACAAATCTGAAGG CACTTTTAGAAAAGTTGCCCACAATTAGTCAGTATGCTGCATTCTTACCTATC ACACCTTCAGAGTTTAGATATATGAATTGTGAAATAAGCAGGGTGATTCTTCC AGAATTGTTATCCATTCATCTTCCTTCATATCCGCCTTACTACCATGTAAGATA TGAAGAAATTGGAGGTGTGGTAAATAATATTCCACATCCAATTGCATAA SEQIDN018; LlsD MISIQNNLEYNKLRWETLSGNVTGIWENNKFFLGSSSYPIMKYHYITANFVNFEK HISENMPKISYHLSGYGVNFNEALVSFIGESAERYTYSLLPT1IKDRIIFRSYEEMTK EYKTDLICELKYINSYYSSEVCENYVTPNDTIQWIAMNSLVHSDKKVWMPLQFV TMYTEEMFSNEKRYVTSAVSTGTACHETVEKSIENALIEYLQIDSFNLWWYGGF RARDIE1DITRNISSWFDNQVAVKKFLSKFNVHFSDISFDKSIYIVLCE1EAKNSSDA FPKYTVGVQGGYSLDKSIYRAFMECLTVLEYNMNVTWTDKEKFLSVTQETRVID NLDDNVIYYSKYGKPELQYNTNQLKNDTEKVTNLKALLEKLPTISQYAAFLPITP SEFRYMNCEISRV1LPELLSIHLPSYPPYYHVRYEE1GGVVNN1PHPIA SEQ ID NO 19; IlsP TTGCATAACATCTTTTTTTTAATCACATTATTTCCAGGTATGCTTCTTTTATTG ACGAAATGGATTCCAGTTCTATCTAGAAAGAGTACTTTTTTTCAATATTTACT TTGTTTATTCTTGATCACAATTATGAATAGTTTGTTTTTCCGTCAACAGTTTGT GGTAGTTTTATCGCTGATATGTATTTTATTCTTACCATTTATTCTGTTTTTTGTA GAATATATATTTGTTGAGAGACAATGGAAAAAGTTGCTTACTATTTATAAAA AAAATAAAATTTTTATCCAATCTATTGTATGGTTTCCTGTTTTAGAAGAAATA ATTTTCCGTTTTTTTATTTATCAATACTGTGAGTTATTTGATTTCAGTAATATC CAGTATATATTACTAGCCACCTTTTCATTCGTGATTGCACATATTTTTTATCAA GGAGTGTCTTCAATTGTCAAAATACTATTTTCTTTTATATTAAGTATATTATTT TTATTAACACTAAATATATTTTTGACAATAATAATTCACTGTATTTTCAACTTT TTAGTTTATATAGTTCGTACTAGTAAATATGAGAACCACCGTAATTGGTAA SEQ ID NO 20; LlsP MHNIFFL1TLFPGMLLLLTKWIPVLSRKSTFFQYLLCLFL1TIMNSLFFRQQFVVVL SLICILFLPFILFFVEYIFVERQWKKLLTIYKKNKIFIQSIVWFPVLEEIIFRFFIYQYC ELFDFSNIQYILLATFSFVIAHIFYQGVSSIVKILFSFILSILFLLTLNIFLTIIIHCIFNFL VYIVRTSKYENHRNW The promoter, PllsA, is located downstream of lmof2365Jlll and it does not contain any of the motifs such as PrfA boxes or σΒ binding sites, previously associated with virulence gene regulation in other L.monocytogenes strains. LlsA encodes the cytotoxin of the nucleic acid sequence of SEQ ID No. 1. The pathogenicity island shares homology with genes associated with the production of previously reported gram-positive bacterium produced cytotoxins, Streptolysin S, Staphylolysin S (Sts) and Botulysin S (Bts) In another aspect the invention provides the cytotoxin is Listeriolysin S (Lis). The cytotoxin has the following charactersitcs: (i) Haemolytic (ii) Cytotoxic IE 0 9q Ο 74 (iii) Contributes to survival of the pathogen in polymorphonuclear neutrophils (PMN). (iv) Contributes to the virulence of the pathogen as assessed by murine assays.
The predicted mass of the leaderless peptide before modification is 2255.7. The peptide is active in a cell-associated form but when in cell-free culture supernatant it is inactive and requires re-activation by the addition of reagents. The re-activation reagent can be RNA core.
The invention further provides a cytotoxin comprising a fragment or a mutant of the cytotoxin of the invention. The invention further provides for probes or antibodies raised against the above-mentioned sequences.
In a further embodiment the invention provides a diagnostic agent to distinguish between high and low virulent strains of L. monocytogenes, based on genes from the LIPI-3 pathogenicity island. The gene may be selected from the genes of thegene region flanked by two Rho-independent terminators, located after Imof2365_ 1111 and llsP, respectively, and two Rho-independent duplicated glyoxalase-encoding genes (lmof2365Jlll, 1121). The genes may be selected from the genes llsA, llsG, llsH, llsX, llsB, llsY, llsD and llsP. The invention also provides a diagnostic kit based on these genes. The agent or kit may comprise e.g. (i) Probes and oligonucleotides for,PCR (both standard and real-time) and hybridisation or assays involving antibodies raised against the cytotoxin or other Lis proteins, (ii) blood agar based assays involving media-condition whereby Lis production is induced while hemolysin (LLO) is repressed (iii) Cell culture based assays etc , although they may form part of many other assays and kits.
The invention also provides for a pharmaceutical composition comprising the cytotoxin of the invention as a means of treatment of a disease by administering a pharmaceutically effective amount of the cytotoxin.
In a still further aspect the invention provides a disinfection agent comprising the cytotoxin defoned above.
The invention also provides the use of Lis in vaccine development. The invention also provides the use of the biosynthetic machinery (i.e. other Lis proteins) involved in the post-translational modification of Lis to post-translationally modify other peptides in a similar manner.
IE Ο 9 ο ο ? 4 LlsA encodes a peptide, which the present inventors have designated Listeriolysin S (Lis). The 49 amino acid pre-propeptide encoded by UsA, resembles the unmodified SagA peptide through the presence of a leader region (26 amino acid) that terminates with a classical ‘double glycine’ bacteriocin-associated cleavage motif (AG) and a putative propeptide (23 aa) with a predominance of cysteine, serine and threonine residues (78%). While the way in which SagA becomes post-translationally modified, it is known that related peptides such as microcin B17 (Mcb) and goadsporin (9, /0), a bacteriocin and a morphogenetic factor, respectively, are modified through the formation of thiazole and oxazole residues (or heterocycles) (//, 12). These modifications require the presence of an associated biosynthetic protein such as McbC (microcin BI7) or, most likely, GodE (goadsporin). LlsB shares homology with these proteins and is thus a member of the McbC-like oxidoreductases, which modify polypeptides by cyclizing thioesters to form rings. It is thus highly likely that Lis is also modified to incorporate heterocycle residues. Mutation of the corresponding Slsassociated gene sagB resulted in the elimination of haemolytic activity (//).
Based on study of the corresponding Group A Streptococcus pathogenicity island, it is highly likely that the majority (and possibly all) of the genes within LIPI-3 are required for Lis production (//). This theory is also supported by the observation that deletion of representative selection of the LIPI-3 genes (i.e. llsH, llsX, llsB, UsY and llsD) from a constitutive Lis producer (F2365LlsCAhly) results in the elimination of haemolytic activity. In contrast deletion of llsP from this background does not result in the elimination of haemolytic activity. It is unclear whether this is as a result of the noninvolvement of llsP or because the need for llsP is somehow overcome as a consequence of expression from a strong constitutive promoter.
In addition to the predicted roles of LlsA and LlsB, as described above, the roles of a number of other LIPI-3 gene products can also be predicted on the basis of bioinformatic analysis. LIsGH represent the individual components of an ABC transporter (22.4% and 19.8% identical to SagGH, respectively). With respect to bacteriocin-like peptides, ABC transporters are most typically associated with export of the structural peptides or are involved in providing protection to the producing strain against the activities of the peptide generated (13, 14). Given that Lis does not exhibit antimicrobial activity, it is most likely that LIsGH is involved in peptide export. Mutation of the corresponding SIs17 £ 09ο 0/4 associated gene sagG resulted in the elimination of haemolytic activity (//) and we have established that deletion of llsH eliminates haemolytic activity from F2365LlsCAA/y.
Given that both components of a two component ABC-transporter are required for functionality, it can be inferred that mutation of llsG would have a similarly detrimental impact.
The role of LlsD (24.7% identity with SagD) is unknown. It has been established that mutation of sagD results in the elimination of Sis activity {Datta, 2005 #370}. We have established that deletion of llsD also eliminates the haemolytic activity of F2365LlsCAWy.
LlsP has been annotated as an N-terminal protease. In a modified bacteriocin context, Nterminal proteases are most frequently associated with bacteriocin leader cleavage i.e. removal of the leader en route to the release of the active portion of the peptide (14). Given their location within this region llsX and llsY it was predicted that they too would play a key role but these proteins do not share homology with proteins of known function and thus it is not possible to predict their role by m-silico analysis alone. We have established that deletion of either of these eliminates the haemolytic activity of F2365LlsCAWy Detailed Description of the Invention The invention will now be described in greater detail with reference to the following figures: Fig. 1. Arrangement of the Stapholysin S (sts), Listeriolysin S (Ils), Streptolysin S (sag) and Botulysin (bts) associated genes in Staph, aureus ET3-1, L. monocytogenes F2365, Strep, pyogenes MGAS 8232 and Cl. botulinum ATCC3502 respectively. Homology between the predicted ils gene products and their sag and sts equivalents is indicated by a line and dashed line, respectively, with an associated % identity value. % identity values with respect to the predicted Sag and Bts gene products are also indicated. The amino acid sequence of the predicted unmodified structural peptides, StsA, LlsA, SagA and BtsA is presented (boxed) with italicized letters indicating amino acids likely to constitute the leader region. Residues within the structural propeptide that are potentially modified are underlined. « 0 9ο 0 7^ Fig. 2. Comparison of the LIPI-3-containing region (bottom) with the corresponding region of Lis' L. monocytogenes (middle), L. innocua and L. welshimeri (top). For L. monocytognes strains, the strain name and serotype (in brackets) is presented and the designation of the first and last gene in each case is that designated in the corresponding genome sequence. Homologous genes (or in the case of the llsA-P, clusters of genes) are presented by matching colours.
Fig. 3. Induction of PllsA by 13mM cumene hydroperoxide and 22 and 50mM hydrogen peroxide. Data are presented as (A.) mean relative light units (RLU; photons s’1) ± standard deviations for three replicates, and (B.) one representative of three independent experiments is shown. The colour bar indicates bioluminescence signal intensity (in photons s'1 cm’2).
Fig. 4. Consequences of constitutive expression or deletion of Ils genes. A. Haemolysis of Columbia blood agar (5% Sheep’s blood) by F2365, F2365A/z/y and F2365EhlyEllsB when Lis is under the control of the natural PllsA or constitutive Phelp promoters (F2365Llsc, F2365LlscAA/y and F2365LlscAWyA//.y#), In situations where haemolytic activity, the corresponding haemoltyic unit (H.U.) values are presented. B. Cytotoxicity relative to F2365 (100%) with respect to the J774, C2-Bbe and CT26 cell lines; F2365Ahly - black, F2365LlsCAA/y - white, * - significantly different (P<0.05), ** extremely significantly different (P<0.005) different. Error bars represent standard error of the mean.
Fig. 5. Comparison of the virulence of F2365 and F2365A//s7S. A. Levels (cfu - colony forming units) of wild-type and mutant F2365 in livers and spleens of Balb/C mice 3 days post-intraperitoneal infection. B. Survival in human PMNs after 2 hr. * - significantly different (P<0.05), ** - extremely significantly different (P<0.005) different. Error bars represent standard error of the mean.
Materials and Methods Growth conditions Listeria were grown in brain heart infusion (BHI) broth or agar (Oxoid) or Columbia blood agar plates (containing 0.5% sheep blood; LIP diagnostics, Galway, Ireland) at 37°C unless otherwise stated. Strains used are listed in tables 1 and 2. Escherichia coli EC 101 (/5) and TOP 10 (Invitrogen) were used as intermediate vector hosts, Antibiotics ΙΕ 09ο 0 were incorporated as follows: Erythromycin (ery), 150pg/ml E. coli, 5pg/ml L. monocytogenes·, Chloramphenicol (cm), 10pg/ml E. coli and L. monocytogenes·, Kanamycin (kan) 50pg/ml E. coli, 25pg/ml L. monocytogenes·, Ampicillin (amp) 100pg/ml E. coli. 5-bromo-4-chloro-3-indolyl-b-D-gaIactopyranoside (X-Gal) was incorporated at a concentration of 40pg/ml.
Strain variation studies For strains of unknown lineage, lineage was determined by an allele-specific oligonucleotide PCR multiplex as described previously (5). LIPI-3 status of strains was determined and confirmed through three distinct PCR reactions involving the primer pairs 1118f-1118r (all primers are listed 5’ to 3’; TCTTACCCTATTATGAAGTATCATAACACCATATCCACTTAAATG), 1118degf-l 1118degr (GATATGTAACTAGCGC TGTATCAACNGGNACNGCT-CCCTCCATACCACCATAAATTAAAKGARTCDAT YTG) and UsAint-llsAsoeD (TGCAGCTGGATGTTGCTC - TAGCCCGGGCAGAACT AAAGT). Amplifications were performed in 50μ1 volumes with 150ng/pl concentrations of each primer, 2mM MgCh, 0.2 mM concentrations of each deoxynucleotide triphosphate, 0.5 U of Taq Polymerase, and 100 ng of genomic DNA. Amplifications consisted of 30 cycles of 1 min at 94°C, 1 min at 56°C, and 1 min at 72°C. Amplification products were resolved on 1.5% (wt/vol) agarose gel.
Genomic Dissimilarity Compositional bias of dinucleotide frequency analysis using the web-based application deltarho (http://deltarho.amc.uva.nl). Deltarho calculates the genomic dissimilarity values d_ (the average dinucleotide relative abundance difference) between input sequences and the genome sequence of choice (16). A high genomic dissimilarity (d_) between an input sequence and the corresponding host genome sequence indicates a heterologous origin of the input sequence.
Investigation of PllsA expression with a luciferase-based reporter system The promoter reporter vector pPLK2-lux was generated by amplifying the kanamycin resistance cassette from pTVl-OK (17) with the primer pair KanRF-KanRR (CCCTGCAGGTCGATAAACC-ACGAATTCCTCGTAGGCGC) and the introduction of the Ry/I-ZlcoRI digested product into similarly-digested pPL2-lux (18). PllsA was fused with the lux cassette within pPLK2-lux through amplification with PllsAfor20 IE 09 Ο Ο 74 PllsAbluntrev (ATTCGTCGACTTTTGATGCTTAAG-CATTCAAATGCCTCCTTTTT ΑΤΤΤ) and cloning into the Sal\-Swa\ sites. The resultant plasmid was isolated from the intermediate host and introduced into F2365. Bioluminescence was investigated by washing overnight cultures and resuspending in an equivalent volume of spent BHI or BHI containing cumene hydrogen peroxide (13 mM) or hydrogen peroxide (22 or 50 mM) and quantified with a Xenogen I VIS 100 imager (Xenogen, Alameda, CA) with a 5 min exposure time.
Constitutive expression of Lis To place the Ils genes under the control of the strong constitutive synthetic promoter Phelp, Phelp DNA was amplified with the primer pair PHELPFsoe-PHELPRsoe (GTGGAGTGAAATATAAGTTAGAGG-TCGAGATCTGCAGATGATTGTGATTTA ATATTCATGGGTTTCACTCTC) from plasmid pPL21uxPhelp (19) and fused between two DNA fragments amplified from the regions flanking PllsA with the primer pairs PllsAchangeA-PllsAchangeB (ACCTGCAGAAGGGGTTATTGA-CTCTAACTTATAT TTCACTCCAC) and PlIsAchangeC-PllsAchangeD (ATGAATATTAAATCACAATCA TC-TGGAATTCCCAGCTCCATTGTCTC) by SOE (splicing by overlap extension) PCR (20) and cloned into the RepA- shuttle vector pORI280 (21) in the intermediate RepA+ host, Escherichia coli EC 101 (/5). The resultant pORI280-help vector was introduced into F2365 already containing the RepA+, temperature sensitive helper plasmid pVE6007 (22). Successfully transformed cells appeared as blue colonies following plating on BHI-Ery-Xgal agar at 30°C. To select for integrants, i.e. cells in which the pORI280-help vector had integrated into the F2365 genome by single crossover homologous recombination, F2365 pVE6007 pOR1280-help was grown in BHI-Ery at 30°C, subcultured twice (0.1% inoculum) in BHI-Ery at 42°C and streaked onto BHI-Ery-Xgal at 42°C. The introduction of Phelp upstream of UsA in EryR/Cms colonies was confirmed by PCR.
Deletion mutagenesis by double crossover homologous recombination The hly gene of F2365 was deleted through the amplification (using the primer pairs hlysoeA-soeB (TGGAATTCCACCTAATGGGAAAGT-GGGTTTCACTCTCCTTCT ACA) and hlysoeC-soeD (TGTAGAAGGAGAGTGAAACCCTAGTGTAGATAATCCAAGCCCGGGACAACTAATCTGAC), splicing and cloning (into the temperature ίΕ 09q sensitive shuttle vector pKSV7(23)) of DNA flanking the gene followed by the introduction of the spliced product through double crossover homologous recombination as described previously (24). This strategy in combination with the primer pairs, HsBsoeA-soeB (ATTCTAGACAAGGTATAGAAAGGTTTGCTGTTTCCTTTCTATGTCTG) and IlsBsoeC-soeD (CAGACATAGAAAGGAAACAGCAAACAAATATTTGTG-TCTCC CGGGAAATAGCTCTTCAC) was also utilized to delete llsB from F2365 and F2365LlscAWy.
Haemolytic assays Haemolysis was assessed by spotting 1 ΟμΙ of overnight cultures onto Columbia blood agar plates and incubating for 24 hr at 37°C. Haemolytic titre assays were carried out as described by Ginsburg et al (Ginsburg et al. JEM 1965), with some minor modification. Washed Listeria were concentrated 10 fold i.e. to 2 χ IO10 cfu/ml and serially diluted (twofold) in a volume of 0.3ml of activation buffer (0.005M maltose, 0.001M MgSCfi7H20,0.001M cysteine) and incubated for 10 min at 37°C.), 0.5ml of prewarmed sheep’s red blood cells (0.2% in activation buffer) was added and the final volume adjusted to 1.0ml. The tubes were incubated at 37°C for 4 hr, centrifuged and haemolysis was assessed spectrophotometrically with a Softmax Pro spectrophotometer at 420 nm. Haemolytic units were calculated for each strain by taking the inverse of the last dilution to show complete haemolysis.
Cytotoxicity assays C2Bbel (CRC-2102; American Type Culture Collection) and J774 (ATCC TIB-67) cells were used for cytotoxicity assays. The cells were maintained in Dulbecco’s modified Eagle’s medium (DMEM) containing 4.5 g/liter Glutamax (Gibco Laboratories, Grand Island, NY), 10% fetal bovine serum (Gibco), 1% (vol/vol) nonessential amino acids (Gibco), 1% (vol/vol) penicillin-streptomycin (Gibco), and 0.01 mg/ml human transferrin (Calbiochem) at 37°C in a 5% CO2 atmosphere. For cell invasion assays, C2Bbe 1 cells were trypsinized (Gibco), harvested by centrifugation at 400 x g for 8 min, and resuspended in 1 ml antibiotic-free DMEM containing 10% fetal bovine serum. Cells were seeded onto 24-well flat-bottom tissue culture plates (Sarstedt, Leicester, United Kingdom) at a concentration of 3 x 105 cells per well. The plates were incubated for 72 h at 37°C in a 5% CO2 atmosphere until confluence was reached. Overnight cultures of L.
IE Ο 9 ο 0 74 monocytogenes were washed and added at a multiplicity of infection of 100:1, and the plates were incubated at 37°C in a 5% CO2 atmosphere for 6 hrs at which time cell-free supernatant was collected and cytoxicity assayed with the Cytotox 96 Non-Radioactive cytotoxicity assay (Promega, Madison, WI) according to the kit instructions.
Murine virulence assay Groups (N=5) of 15-week-old BALB/c mice were inoculated intraperitoneally with overnight cultures of F2365 or F2365kllsB resuspended in 0.2 ml of phosphate-buffered saline to a final concentration of 2 x 106 CFU/ml. Mice were sacrificed 3 days postinfection, and the numbers of L. monocytogenes in the livers and spleens of infected animals were determined by plating serial 10-fold dilutions of organ homogenates on BHI agar. All procedures involving the use of animals were approved by the institutional animal care committee and complied with relevant legal guidelines.
Isolation of human PMNs Polymorphonuclear neutrophil granulocytes (PMNs) were isolated by a one step procedure based on a method described by English and Andersen (25). Briefly, 9 ml EDTA anticoagulated peripheral blood obtained from healthy donors was diluted with an equal volume of 0.9% NaCI and carefully overlain on a discontinuous double gradient formed by layering 4 ml of polysucrose/sodium diatrizoate adjusted to a density of 1.077 g/ml (Histopaque 1077) on 4 ml Histopaque 1119 in 15 ml conical centrifuge tubes (Starstedt UK). The tubes were subsequently centrifuged at 700 x g for 30min at room temperature. After centrifugation two distinct leukocyte cell layers (lymphocytes/monocytes and PMNs, respectively) were obtained above the bottom sediment of erythrocytes. The PMN layer was carefully aspirated and washed in DMEM supplemented with 10% fetal calf serum (DMEM-FCS). Centrifugation followed at 300 x g for 10 min at room temperature. After two further washes the cells were counted using trypan blue staining solution (Sigma-Aldrich Chemie, Deisenhofen, Germany) and the viability of the cells was confirmed to be above 97%.
Quantification of intracellular survival of L. monocytogenes in human PMNs.
Human PMNs were isolated as described above and were adjusted to a final concentration of 1 x 106 cells/ml in DMEM-FCS. Overnight cultures of L. monocytogenes were washed with PBS, and resuspended in DMEM-FCS at a final *Ε Ο 9 ο 074 concentration of 2 χ lO7/mL. PMNs and L. monocytogenes were combined to yield a final bacteria:PMN ratio of 20:1. Incubation was at 37°C in 5% CO2. After 30 mins the PMNs were washed, resuspended in fresh medium containing 50 pg/ml of gentamicin (Sigma) to kill extracellular bacteria, and further incubated until T=2hrs. Subsequently, the samples were washed with PBS, and cells were lysed by the addition of ice-cold water, serially diluted in PBS, and plated on BHI agar to determine the number of viable intracellular bacteria.
Statistical analysis In all cases the differences in mean values were analysed with an independent samples ttest, following testing for conformity of data to assumptions of parametric statistics (Kolmogorov-Smirnov test for normality and Levene’s test for equality of variances). Results BLAST Sequence Comparisons DNA and protein databases were screened, utilizing the NCBI sequence database, with respect to genes associated with the production of bacteriocins or bacteriocin like peptides with particular interest in sagB/SagB homologues. This revealed that lineage IL. monocytogenes strains, F2365 and H7858, possess genes that are homologous to SagB (24% identity). SagB is a protein associated with the formation of Streptolysin Sis. These homologues are situated within an 8-gene island, provisionally named LIPI-3 by the current inventors (Figure 1).
Bioinformatic analysis of LIPI-3 revealed the presence of several additional genes with homology to Streptolysin-associated genes (sag). The sag locus, which includes nine genes (sag A-sag I), is necessary and sufficient for the production of Streptolysin S. The sag genes consist of sagA (encoding a 73% Cys/Gly/Ser/Thr-rich structural propeptide with a bacteriocin-like leader), sagB (a putative modification protein containing a McbClike oxidoreductase domain, cd02142.1), sagE (a putative immunity protein), sagGHIfpn ABC transporter potentially involved in export) as well as sagC, D and A (encoding proteins of unknown function).
The ORFs designated llsGHBDP encode LlsGH, both components of an ABC transporter (22.4% and 19.8% identical to SagGH, respectively), LlsB (24.7% identity with SagB), LlsD (24.7% with SagD) and LlsP has been annotated as an N-terminal protease fE Ο 9 ο ο 7 (frequently associated with bacteriocin leader cleavage). Given their location within this region llsX and UsY could also be of relevance. This is further illustrated in Fig I.
The Sls-like features of LIPI-3 include the presence of a previously un-annotated putative structural gene designated by the current inventors as llsA. LlsA is predicted to encode a peptide, which the present inventors have designated Listeriolysin S (Lis). The 49 amino acid pre-propeptide encoded by llsA, resembles the unmodified SagA peptide through the presence of a leader region (26 amino acid) that terpiinates with a classical ‘double glycine’ bacteriocin-associated cleavage motif (AG) and a putative propeptide (23 aa) with a predominance of cysteine, serine and threonine residues (78%) (Figure 1).
LIPI-3 Detection PCR was utilised to determine the strain variable nature of LIPI-3 (the 8 gene island between the 2 extreme rho-independent terminators). Listeria isolates representing a cross-section of the three lineages as well as non-Lmonocytogenes strains. A number of PCR primers were utilised which targeted different regions of the island. 1118f — 1118r and 1118degF-l 118degR both amplify a stretch within HsD while UsAint-llsAsoeD amplify from the middle of llsA into llsG.
LIPI-3 was not detected in any of the lineage II and lineage III L.monocytogenes strains tested or in any non L. monocytogeneous strains tested i.e. L. innocua, L.welshimeri, L. seeleri, L. ivanovii and L. grayi. 52% of lineage I strains tested were found to contain LIPI-3 (Table 1, Table 2). The LIPI-3+ strains corresponded to 13 of the 19 lineage I )· sequence types (STs) previously proposed, i.e. STs 3-10,13-14 and 17-19. Furthermore, in addition to F2365 and H7858, responsible for epidemic outbreaks of listeriosis in California (1985) and the US (multistate; 1998-99), respectively, the LIPI-3+ set includes the listeriosis-outbreak strains from Halifax (1981), Lausanne (1987), Illinois (1994) and North Carolina (2000). In contrast, relatively few lineage I outbreak-associated strains (i.e. Massachusetts 1985, UK 1989) lack LIPI-3+ In Silico Analysis and Screening In silico analysis established that in contrast to LIPI-1, which was present in a common listerial ancestor but then lost from non-pathogenic species (26), the very high genomic dissimilarity value of LIPI-3 (δ* of 0.1178) relative to that of the remainder of the F2365 genome (δ* of 0.0343) and its atypical %GC content (29.9 vs 38%; Table SI) indicate that it has been acquired relatively recently. Interestingly, this region of the listerial genome seems to be particularly variable in that H7858 possesses an additional 14 ORFs upstream of its lmof2365_Jlll equivalent and there is a large amount of inserted DNA (> 17kB) at the corresponding location in EGDe (Fig. 2), both of which are absent from F6854, L. innocua CLIP 11262 and L. welshimeri SLCC5334, Analysis of the corresponding regions from recently released whole genome shotgun sequence data of a number of L. monocytogenes strains generated by the Broad Institute emphasises this point (27); Fig. 2. Such analysis also reveals that homologs of lmof2365 1111 and lmofl365_1120 are to be found in LIPI-3” strains, thereby confirming that the flanking rho-independent terminators mark the outer limits of LIPI-3 (Fig. 2). In silico screening also revealed that, in addition to specific L. monocytogenes, GAS, related group C and G streptococci Streptococcus iniae (28), other genome-sequenced Gram positive pathogens i.e. Staphylococcus aureus ET3-1, a representative of the most abundant bovine mastitisassociated Staphylococcus aureus lineages, and a number of group I Clostridium botulinum strains, possess clusters of related open reading frames (ORFs). Although the existence of these islands has also been noted recently by others (29, 30), they have not been described in great detail. The putative products of these ORFs have been designated Staphylolysin S (Sts) and Botulysin S (Bts).
StsA encoded within this cluster is predicted to possess a leader (26 aa), leader cleavage site (AG), a structural propeptide (24 aa) with a large number of potentially modified residues (83% Cys/Ser/Thr) and an N-terminal CCSCSCS motif also present in LlsA (Figure 1).
Gene arrangement and % identity values established that the Lis (LIPI-3) and Sts clusters are more closely related to one another than to their Sis counterpart. In fact of the eight genes located on LIPI-3, only llsX does not have significant identity with its Sts equivalent. Notably however, within the Sts cluster, homology to both llsB and llsD is divided across two ORFs. Resequencing of this region has confirmed these to be genuine frameshifts.
The Bts gene cluster was found to closely resemble Sis gene cluster at a region predicted to encode a structural propeptide (33 aas) having up to 73% modified residues (Fig.l), Haemolytic assays ΙΕ ο 9 ο 0 74 Sis is responsible for the haemolytic activity of GAS. As a result the haemolytic activity of LIPI-3 was investigated to determine whether it was responsible for the production of a haemolytic factor. As all L. monocytogenes strains, regardless of lineage, produce the cytolysin listeriolysin 0 (encoded by hly located within LIPI-1) (31-34) which could mask any Lis activity, a F2365AWy mutant was created. The Bhly mutant was nonhaemolytic when grown on Columbia blood agar. To determine whether the Ils genes are expressed under such conditions PllsA was fused to a Lux reporter system in a promoter probe vector (pPLKm2) and integrated as a single copy into the F2365 genome. Realtime analysis with an I VIS 100 imager (Xenogen) revealed that expression under routine laboratory growth conditions is negligible but that the promoter was strongly induced upon exposure to oxidative stress, including cumene hydrogen peroxide (13 mM) and hydrogen peroxide (22 and 50 mM) for 10 min (Fig. 3).
The induction is transient, dissipating gradually over a 60 min period. The transient nature of this induction, combined with the haemolytic nature of cumene hydrogen peroxide at these concentrations, restricted assessment of the consequences of expression of the Lis genes from a haemolytic perspective. To overcome this problem the Ils genes were placed under the control of a constitutively strong synthetic Gram positive promoter Phelp in both F2365 and F2365A/rZy such that the consequences of Lis production could be determined. Placing the genes under the control of Phelp enhanced the haemolytic activity of F2365 (F2365Lls ) and resulted in a haemolytic phenotype in the Ahly background (i.e. F2365Lls Ahly) (Fig. 4a). This haemolytic activity was eliminated when llsB was mutated in a non-polar fashion, confirming the link between the island and haemolysis (Fig. 4a). L. monocytogenes cells were employed for these, and other phenotypic assays, as it was apparent that Lis, like Sis, is active in a cell-associated form with no activity being apparent from untreated cell-free culture supernatant. Data analogous to that in fig 4c demonstrates that the constitutive Lis producing strain has activity against the mouse colon carcinoma cell line CT26.
Cytotoxicity Assays Γ' The cytotoxicity of the Lis gene cluster was also assessed using F2365Lis Ahly, F2365BhlyisllsB and a number of different cell lines. F2365LlscAWy was significantly Ί 4 more cytotoxic than F2365A/z/y against C2-Bbe (human enterocyte-like), J774 (mouse macrophage) and CT26 (mouse colon carcinoma) cell lines (Fig. 4b).
Murine virulence studies The contribution of LIPI-3 to pathogenicity was assessed by comparing the virulence of F2365 and F2365A/kS (//<) following intraperitoneal inoculation of Balb/c mice. From these assays it was apparent that Ils possessed a reduced virulence potential as evidenced by significantly and extremely significantly reduced levels in the livers and spleens, respectively, relative to the corresponding F2365 infected mice (Fig. 5a).
PMN Survival A number of lines of evidence suggested that one of the roles of Lis may be to contribute to the survival of L. monocytogenes in PMNs. Firstly, Sis contributes to the ability of GAS to withstand neutrophil killing (//); secondly, of the phagocytes, PMNs produce the greatest concentration of oxidative stress-inducing species; and thirdly, the initial response to intraperitoneal infection involves an influx of neutrophils). Notably, PMNs are essential for the resolution of L. monocytogenes infections, playing a critical role in reducing the bacterial burden in the liver, spleen and central nervous system (35). When the intracellular survival of F2365 and Ils in purified human PMNs (human cells being used to establish the human relevance of the murine results) was compared after 2 hrs, it was apparent that wild-type F2365 again survived significantly better than the Ils mutant (Fig. 5b), confirming a role for Lis as a virulence factor mitigating survival in PMNs. Discussion The current inventors have identified an island of genes, provisionally named LIPI-3, within the genome of a subset of strains of L. monocytogenes. Regions within this island were found to be homologous to regions within the genome of Group A Streptococcus (GAS), S. aureus and Cl. Boutulinum. The regions of homology were those responsible for the biosynthesis of the highly cytotoxic Group A Streptococcus (GAS) virulence factor Streptolysin S, the S. aureus associated Staphylolysin S (Sts) and the Cl.
Boutulinum associated Botulysin S (Bts). Previous studies have reported that Streptolysin S is responsible for the characteristic β-haemolytic activity of GAS and plays a role in the survival of GAS in polymorphonuclear neutrophils (PMNs), cytotoxicity and inflammatory activation, contributing to necrosis and systemic spread.
It has been assumed throughout the literature that the thiol-activated cytotoxin listeriolysin 0 (LLO) was the sole haemolysin/cytolysin produced by L.monocytogenes strains. In the process of disputing this, the current inventors have established that LIPI-3 is specifically associated with a subset of lineage I strains. To access the strain variable nature of LIPI-3, Listeria isolates representing a cross-section of the three lineages as well as non-monocytogenes species were tested. 52% of lineage I strains tested were found to contain LIPI-3. In addition to F2365 and H7858, responsible for epidemic outbreaks of listeriosis in California (1985) and the US (1998-1999) the LIPI-3+ set included listeriosis-outbreak strains from Halifax (1981), Lausanne (1987), Illinois (1994) and North Carolina (2000). No lineage II or lineage II strains tested or any of the non-monocytogenes strains tested were found to contain the island LIPI-3. LIPI-3 contains 8 genes including a previously unannotated gene UsA. LlsA encodes a cytolytic virulence factor designated Listeriolysin S. In the present study the current inventors have shown that Listeriolysin S has the following characteristics: (i) Haemolytic (ii) Cytotoxic (iii) Contributes to survival of the pathogen in polymorphonuclear neutrophils (PMN).
The predicted mass of the leaderless peptide before modification is 2255.7. The peptide is active in a cell-associated form but when in cell-free culture supernatant it is inactive and requires re-activation by the addition of reagents such as RNA core.
These findings, coupled with the contribution of listeriolysin S at a critical junction during the pathogenic life of this pathogen, point to Lis as being a cytotoxin associated with the high virulence potential of lineage I strains.
Currently there are no efficient or reliable diagnostic tests available to distinguish between highly virulent and low virulent strains of L.monocytogenes. The current invention provides a solution to this by providing factor specific for a subset of lineage I strains that can be used in a diagnostic test as a marker of high virulence. Should it be accepted by regulatory agencies that LIPI-3 is indeed responsible for the apparent enhanced virulence potential of some strains of L.monocytogenes, it may be that the limits with respect to the quantities of L.monocytogenes that are permitted in foods will IE Ο 9 ο ο be altered such that there are two limits. A low limit for the more virulent LIPI-3+ strains and a higher limit for LIPI-3- strains. Methods for the detection of LIPI-3 in L.monocytogenes strains could involve conventional PCR, Real-Time PCR or DNA hybridizations.
Furthermore, listeriolysin S has the potential application to function as a chemotherapeutic agent, acting as an alternative to existing agents. The current study has shown that listeriolysin S is cytotoxic to a number of cell types including cancerous cells. •Ε 0 9 0 0 74 Table 1.
Lis status of non-lineage I Listeria Lineage* Strain1 Equivalent Original source Serotype Lis1 L. monocytogenes 1IA 33225ac LMB0455 3a I IIA 33226ac LMB0456 3c 1 11B NCTC7973 Clinical l/2a [ IIs CD1061 Pork sausage non 4 J IIA 33022ac ATCC 15313 Rabbit l/2a 1 IIs DPC4605 SLCC2479 Unknown 3c _1 1IB CD1198 ground turkey non 4 J IIs CD1038 Pork sausage non 4 ) IIB CD1059 Pork sausage 1 2 I IIs CD241 Silage 3 J IIs CD1O28 Pork sausage non-4 1 IIB CD243 Silage 1 /2 J IIs CD 1742 Pork sausage non-4 J IIA EGDe laboratory strain l/2a 1,2 IIA 33234a F6854 turkey frankfurter l/2a 2 1IA I0403S laboratory strain l/2a 2 I1A J2818 turkey deli (US 1989) l/2a 2 IIA FSLN3-165 Soil l/2a _2 I1A FSL Jl-101 33418, F6900 Clinical l/2a _2 irA LO28 laboratory strain l/2c 2 I1A FSLJ2-003 l/2a _2 1IA J0161 33419, FSL R2499 turkey deli (US 2000) l/2a 2 I1A FSLF2-515 l/2a 2 1I1A 33077ac 98-18140 bovine tissue 4b 1I1A 33115ac 93-500 Arabia oryx 4c _i II1B CD83 Silage 4 1 1IIA FSLJ2-071 4c 2 111A FSL J1-208 animal clinical 4a _2 L. innocua CLIP 11262 ,2 FH2333 Lettuce J FH2381 Sausage rolls 1 L. ivatiovii CD293 _1 CD588 1 CD165 L. grayi ATCC25403 Corn stalks 1 ATCC25400 Corn stalks _1 FH2289 Cooked chicken J L. welshimeri SLCC5334 2 1312109 Chicken J FH1968 Caesar salad 1 IE 0 9 0 0 74 ATCC35897 Plant material /.. seelegeri CD 166 FH2O62 Cooked breakfast CD944 Table 2 Lis status of lineage IL. monocytogenes Lineage* Strain1 Equivalent Source ST* Serotype LIs* IA 330l3ac ScottA Clinical (Mass, outbreak, 1983) 1-la 4b IA 33413ac Ts45 Food (UK outbreak, 1988) Mb 4b IA 33007ac RM2218 Food l-2a 4b IA 33008ac RM2387 Food l-3a 4b +1 1A 33083ac FI 109 Food 1 -4 a 4b +’ IA 33420a NI-227, H7738 Food (US outbreak, 199899) l-5a 4b +' 1A 33233ar H7858 Food (US outbreak, 199899) l-5b 4b +!’2 IA 33386ac NI-225, H7550 Clinial (US outbreak, 1998-99) l-5b 4b +' IA 33104a F2365 Ca 1985 1 -6 a 4b +'·2 1A 33410a JI-119, TS43, F.4565 Clinical (L.A. outbreak, 1985) 1 -6 a 4b +' IA 33411a FSLN3-OO8, TS50, L.4760 Food (Halifax outbreak, 1981) l-6b 4b +' 1A 33415a FSLN3-022, TS21, L.4486j Food (Switzerland outbreak, 1987) l-6c 4b +' IA 33120a ATCC19118 Animal l-6c 4e +' IA 33116a ATCC19117 Animal l-7a 4d +' IA 33OI5ac 12375 l-8a 4b +' IA 33424a FSL R2-503 Clinical (Illinois outbreak, 1994) l-9a l/2b +2 lA 33423ac G6003 Food l-9a l/2b +' IA 33068ac 8058 Animal l-10a l/2b +' IA 33038ac OB001385 Food 1-lla l/2b i IA 33037ac OB001350 Food l-12a l/2b 1 IA 33126ac 7034 Animal l-13a l/2b +' 1A 33176ac 20240-954 Animal l-14a l/2b +' IA 33090ac 7675 Animal l-15a l/2b 1 IA 33028ac OBOOl 102 Food l-16a l/2b 1A 33390a FSL J2-064 l-16f l/2b 2 IA 33032ac OBOOl186 Food l-17a l/2b +' IA 33186ac 20674-01 Animal l-18a l/2b +' 1A 33421c JO 144 Food (Nrth Car. outbreak, 2000) l-19a 4b but 1 /2b complex +1 IE 0 9 0 0 74 ΙΛ GDI 121 ground beef Clinical (Mass, outbreak, nd 4b 1 IA F5817 1983) nd non 4 1 1“ CD2088 pork sausage Clinial outbreak (Mass. nd 4b 1 IA FSLJ1-220 outbreak, 1983) nd l/2b +1 IB DPC4608 SLCC1694 Unknown nd non-4 1 IB CD1032 Pork sausage nd 4a J IB CD 147 Dairy enrichment nd non 4 +1 IB CD 1066 Pork sausage nd non-4 1 IB CD749 Ground beef nd non-4 1 IB CD748 Ground beef nd 4 1 IB CD246 Silage nd 4 ] lB CD878 Clinical nd 1 2 +' IB CD1078 Chicken nd l/2b 2 IA FSL Jl-175 Water nd l/2b 2 IA FSL J1-194 Sporadic clinical nd 4b +2 IA FSL Nl-017 Trout in brine nd 4b 2 IA HPB2262 Febrile cases (Italy) nd 4b 2 Table 3 Island 1000 χ δ* island (genomic disimilarity) 1000 χ δ* genome average % GC Island % GC Genome Lis 117.8 34.3 29.9 38 Sts 61.3 45 26.6 32.8 Sag 35.4 36.2 35 38.5 Bts 33.8 33.6 26.2 28.2 IE 0 9 0 0 74 References 1. T. J. Ward et al., JBacteriol 186, 4994 (Aug, 2004). 2. T. Revazishvili et al.,JClm Microbiol 42,276 (Jan, 2004). 3. C. Salcedo, L. Arreaza, B. Alcala, L. de la Fuente, J. A. Vazquez, J Clin Microbiol 41, 757 (Feb, 2003). 4. W. Zhang, Β. M. Jayarao, S. J. Knabel, Appl Environ Microbiol 70,913 (Feb, 2004).
. T. F. Ducey et al.,Appl Environ Microbiol 73, 133 (Jan, 2007). 6. R. Brosch, C. Buchrieser, J. Rocourt, Res Microbiol 142, 667 (Jul-Aug, 1991). 7. A. De Cesare, J. L. Bruce, T. R. Dambaugh, Μ. E. Guerzoni, M. Wiedmann, J Clin Microbiol 39, 3002 (Aug, 2001). 8. K. Grif, 1. Heller, M. Wagner, M. Dierich, R. Wurzner, Foodborne PathogDis 3, 138 (2006). 9. H. Onaka, M. Nakaho, K. Hayashi, Y. Igarashi, T. Furumai, Microbiology 151, 3923 (Dec, 2005).
. Y. M. Li, J. C. Milne, L. L. Madison, R. Kolter, C. T. Walsh, Science 274, 1188 (Nov 15, 1996). 11. V. Datta et al., Mol Microbiol 56, 681 (May, 2005). 12. V. Nizet et al., Infect Immun 68,4245 (Jul, 2000). 13. P. D, Cotter, C. Hill, R. P. Ross, Nat Rev Microbiol 3, ΊΊΊ (Oct, 2005). 14. O. McAuliffe, R. P. Ross, C. Hill, FEMS Microbiol Rev 25, 285 (May, 2001).
. J. Law et al., J Bacteriol 177,7011 (Dec, 1995). 16. M. W. van Passel, A. C. Luyf, A. H. van Kampen, A. Bart, A. van der Ende, Bioinformatics 21,3053 (Jul 1,2005), J. A. Gutierrez et al., J Bacteriol 178, 4166 (Jul, 1996). 18. P. A. Bron, I. R. Monk, S. C. Corr, C. Hill, C. G. Gahan, Appl Environ Microbiol 72, 2876 (Apr, 2006). 19. C. U. Riedel et al., Appl Environ Microbiol 73, 3091 (May, 2007).
. R. M. Horton, Z, L. Cai, S. N. Ho, L. R. Pease, Biotechniques 8, 528 (May, 1990). 21. K. Leenhouts et al., Mol Gen Genet 253, 217 (Nov 27, 1996). 22. E. Maguin, P. Duwat, T. Hege, D. Ehrlich, A. Gruss, J Bacteriol 174, 5633 (1992). 23. K. Smith, P. Youngman, Biochimie 74, 705 (Jul-Aug, 1992).
IE Ο 9 Ο Ο 7 4 24. Ρ. D. Cotter, C. G. Gahan, C. Hill, Mol Microbiol 40, 465 (Apr, 2001).
. D. English, B. R. Andersen, J Immunol Methods 5, 249 (Aug, 1974). 26. T. Hain et al., JBacteriol 188, 7405 (Nov, 2006).
Listeria monocytogenes Sequencing Project. Broad Institute of Harvard and MIT (http://www.broad.mit.edu) 28. V. Nizet, Trends Microbiol 10, 575 (Dec, 2002). 29. L. Herron-Olson, J. R. Fitzgerald, J. M. Musser, V. Kapur, PLoS ONE 2, el 120 (2007).
. M. Sebaihia et al., Genome Res 17, 1082 (Jul, 2007). 31. A. J. Roberts, M. Wiedmann, Cell Mol Life Sci 60,904 (May, 2003). 32. M. Hamon, H. Bierne, P. Cossart, Nat Rev Microbiol 4,423 (Jun, 2006). 33. S. Kayal, A. Charbit, EEMS Microbiol Rev 30, 514 (Jul, 2006). 34. G. Dominguez-Bernal et al., Mol Microbiol 59, 415 (Jan, 2006).
. S. Lopez, A. J. Marco, N. Prats, C. J. Czuprynski, Infect Immun 68,4789 (2000).

Claims (5)

Claims
1. A gene derivable from the LIPI-3 pathogenicity island of the gram positive pathogen Listeria monocytogenes, the island being flanked by two Rho-independent terminators, located after lmof2365_1111 and llsP, respectively, and two Rhoindependent duplicated glyoxalase-encoding genes (Imof2365_llll, 1121),
2. Λ sequence as set forth in SEQ ID No. 1 or 2, in SEQ ID NO 6, 8, 10, 12, 14, 16,18, and 20or a sequence substantially similar thereto which also encodes a cytotoxin or cytotoxic activity.
3. A cytotoxin, Listeriolysin S, wherein the cytotoxin is haemolytic to sheep red blood cells, cytotoxic to C2-Bbel, J774 and CT-26 cells and contributes to survival of the pathogen following polymhorphonuclear neutrophils (PMN) challenge and to virulence, as assessed by murine assays.
4. Use of a gene as claimed in claim 1, a sequence as claimed in claim 2 or a cytotoxin as claimed in claim 3 in a diagnostic kit for distinguishing between high and low virulent strains of Listeria monocytogenes, in a pharmaceutical composition, or in a disinfectant composition.
5. A gene, an antibody raised against a the product of the gene, a nucleic acid sequence, an amino acid sequence, a cytotoxin, a diagnostic kit or a composition substantially as described herein with reference to the Examples.
IES20090074 2008-01-24 2009-01-23 Listeria monocytogenes cytotoxin listeriolysin S IES20090074A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
IES20090074 IES20090074A2 (en) 2008-01-24 2009-01-23 Listeria monocytogenes cytotoxin listeriolysin S

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
IE20080050A IE20080050A1 (en) 2008-01-24 2008-01-24 Listeria monocytogenes cytotoxin Listeriolysin S
IES20090074 IES20090074A2 (en) 2008-01-24 2009-01-23 Listeria monocytogenes cytotoxin listeriolysin S

Publications (1)

Publication Number Publication Date
IES20090074A2 true IES20090074A2 (en) 2009-10-28

Family

ID=40673323

Family Applications (2)

Application Number Title Priority Date Filing Date
IE20080050A IE20080050A1 (en) 2008-01-24 2008-01-24 Listeria monocytogenes cytotoxin Listeriolysin S
IES20090074 IES20090074A2 (en) 2008-01-24 2009-01-23 Listeria monocytogenes cytotoxin listeriolysin S

Family Applications Before (1)

Application Number Title Priority Date Filing Date
IE20080050A IE20080050A1 (en) 2008-01-24 2008-01-24 Listeria monocytogenes cytotoxin Listeriolysin S

Country Status (2)

Country Link
IE (2) IE20080050A1 (en)
WO (1) WO2009092790A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3219721A1 (en) 2016-03-16 2017-09-20 Institut Pasteur Listeriolysin s or related peptides as antibacterials

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999049049A1 (en) * 1998-03-20 1999-09-30 Mount Sinai Hospital Streptococcus sag-a, a structural protein associated with sls activity
FR2814754B1 (en) * 2000-10-04 2005-09-09 Pasteur Institut LISTERIA INNOCUA, GENOME AND APPLICATIONS
US20060257894A1 (en) * 2003-09-16 2006-11-16 Michel Doumith Molecular typing of listeria monocytogenes, hybridization supports and kits for said molecular typing

Also Published As

Publication number Publication date
WO2009092790A1 (en) 2009-07-30
IE20080050A1 (en) 2009-11-11

Similar Documents

Publication Publication Date Title
Cotter et al. Listeriolysin S, a novel peptide haemolysin associated with a subset of lineage I Listeria monocytogenes
O'Shea et al. Production of multiple bacteriocins from a single locus by gastrointestinal strains of Lactobacillus salivarius
JP2801322B2 (en) DNA probes specific for hemolytic Listeria
Musser et al. Genetic diversity and relationships among Streptococcus pyogenes strains expressing serotype M1 protein: recent intercontinental spread of a subclone causing episodes of invasive disease
Mishra et al. Sortase-catalyzed assembly of distinct heteromeric fimbriae in Actinomyces naeslundii
Tegetmeyer et al. ISApl1, a novel insertion element of Actinobacillus pleuropneumoniae, prevents ApxIV-based serological detection of serotype 7 strain AP76
Chimalapati et al. Effects of deletion of the Streptococcus pneumoniae lipoprotein diacylglyceryl transferase gene lgt on ABC transporter function and on growth in vivo
JP2001503970A (en) Novel bacteriocin, transport and vector systems and methods of use thereof
Pérez-Pascual et al. RovS and its associated signaling peptide form a cell-to-cell communication system required for Streptococcus agalactiae pathogenesis
CA2517899A1 (en) Genes of an otitis media isolate of nontypeable haemophilus influenzae
Maricic et al. Characterization of a multipeptide lantibiotic locus in Streptococcus pneumoniae
Knijff et al. Rapid identification of Enterococcus durans and Enterococcus hirae by PCR with primers targeted to the ddl genes
Wu et al. A Streptococcus suis LysM domain surface protein contributes to bacterial virulence
Seto et al. Properties of corynephage attachment site and molecular epidemiology of Corynebacterium ulcerans isolated from humans and animals in Japan
Asam et al. Identification of β‐haemolysin‐encoding genes in S treptococcus anginosus
Dufour et al. Genetic analysis of mutacin B-Ny266, a lantibiotic active against caries pathogens
Matos et al. Study on the dissemination of the bcrABDR cluster in Enterococcus spp. reveals that the BcrAB transporter is sufficient to confer high-level bacitracin resistance
Callahan et al. The Streptococcus sanguinis competence regulon is not required for infective endocarditis virulence in a rabbit model
Lomonaco et al. Analysis of additional virulence genes and virulence gene regions in Listeria monocytogenes confirms the epidemiologic relevance of multi-virulence-locus sequence typing
Bigot et al. A homolog of Bacillus subtilis trigger factor in Listeria monocytogenes is involved in stress tolerance and bacterial virulence
Cooper et al. Re-sequencing of a virulent strain of Campylobacter jejuni NCTC11168 reveals potential virulence factors
Chatterjee et al. Invasiveness is a variable and heterogeneous phenotype in Listeria monocytogenes serotype strains
Kawada-Matsuo et al. Two-component systems involved in susceptibility to nisin A in Streptococcus pyogenes
IES20090074A2 (en) Listeria monocytogenes cytotoxin listeriolysin S
Reimundo et al. dltA gene mutation in the teichoic acids alanylation system of Lactococcus garvieae results in diminished proliferation in its natural host

Legal Events

Date Code Title Description
MM4A Patent lapsed